Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 966
Filter
1.
Stroke ; 53(2): e37-e41, 2022 02.
Article in English | MEDLINE | ID: mdl-34743535

ABSTRACT

BACKGROUND AND PURPOSE: Neuroprotective strategies for stroke remain inadequate. Nanoliposomes comprised of phosphatidylcholine, cholesterol, and monosialogangliosides (nanoliposomes) induced an antioxidant protective response in endothelial cells exposed to amyloid insults. We tested the hypotheses that nanoliposomes will preserve human neuroblastoma (SH-SY5Y) and human brain microvascular endothelial cells viability following oxygen-glucose deprivation (OGD)-reoxygenation and will reduce injury in mice following middle cerebral artery occlusion. METHODS: SH-SY5Y and human brain microvascular endothelial cells were exposed to oxygen-glucose deprivation-reoxygenation (3 hours 0.5%-1% oxygen and glucose-free media followed by 20-hour ambient air/regular media) without or with nanoliposomes (300 µg/mL). Viability was measured (calcein-acetoxymethyl fluorescence) and protein expression of antioxidant proteins HO-1 (heme oxygenase-1), NQO1 (NAD[P]H quinone dehydrogenase 1), and SOD1 (superoxide dismutase 1) were measured by Western blot. C57BL/6J mice were treated with saline (n=8) or nanoliposomes (10 mg/mL lipid, 200 µL, n=7) while undergoing 60-minute middle cerebral artery occlusion followed by reperfusion. Day 2 postinjury neurological impairment score and infarction size were compared. RESULTS: SH-SY5Y and human brain microvascular endothelial cells showed reduced viability post-oxygen-glucose deprivation-reoxygenation that was reversed by nanoliposomes. Nanoliposomes increased protein expressions of HO-1, NQO1 in both cell types and SOD1 in human brain microvascular endothelial cells. Nanoliposomes-treated mice showed reduced neurological impairment and brain infarct size (18.8±2% versus 27.3±2.3%, P=0.017) versus controls. CONCLUSIONS: Nanoliposomes reduced stroke injury in mice subjected to middle cerebral artery occlusion likely through induction of an antioxidant protective response. Nanoliposome is a candidate novel agent for stroke.


Subject(s)
Infarction, Middle Cerebral Artery/drug therapy , Liposomes/therapeutic use , Nanoparticles/therapeutic use , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Animals , Antioxidants/metabolism , Cell Line , Endothelium, Vascular/pathology , Glucose/deficiency , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Humans , Hypoxia , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Microvessels/pathology , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , NAD(P)H Dehydrogenase (Quinone)/genetics , Reperfusion Injury/pathology , Stroke/etiology , Stroke/pathology , Superoxide Dismutase-1/biosynthesis , Superoxide Dismutase-1/genetics
2.
Mol Cell Biol ; 41(11): e0023621, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34398680

ABSTRACT

Heme oxygenase 1 (HO-1) is the key enzyme for heme catabolism and cytoprotection. Whereas HO-1 gene expression in response to various stresses has been investigated extensively, the precise mechanisms by which HO-1 gene expression is regulated by the HO-1 substrate heme remain elusive. To systematically examine whether stress-mediated induction and substrate-mediated induction of HO-1 utilize similar or distinct regulatory pathways, we developed an HO-1-DsRed-knock-in reporter mouse in which the HO-1 gene is floxed by loxP sites and the DsRed gene has been inserted. Myeloid lineage-specific recombination of the floxed locus led to fluorescence derived from expression of the HO-1-DsRed fusion protein in peritoneal macrophages. We also challenged general recombination of the locus and generated mice harboring heterozygous recombinant alleles, which enabled us to monitor HO-1-DsRed expression in the whole body in vivo and ex vivo. HO-1 inducers upregulated HO-1-DsRed expression in myeloid lineage cells isolated from the mice. Notably, analyses of peritoneal macrophages from HO-1-DsRed mice lacking NRF2, a major regulator of the oxidative/electrophilic stress response, led us to identify NRF2-dependent stress response-mediated HO-1 induction and NRF2-independent substrate-mediated HO-1 induction. Thus, the HO-1 gene is subjected to at least two distinct levels of regulation, and the available lines of evidence suggest that substrate induction in peritoneal macrophages is independent of CNC family-based regulation.


Subject(s)
Gene Expression Regulation/genetics , Heme Oxygenase-1/genetics , Heme/metabolism , Macrophages, Peritoneal/metabolism , Membrane Proteins/genetics , Oxidative Stress/genetics , Animals , Antioxidants/metabolism , Bilirubin/metabolism , Biliverdine/metabolism , Heme Oxygenase-1/biosynthesis , Luminescent Proteins/genetics , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Oxidative Stress/physiology
3.
J Biochem Mol Toxicol ; 35(8): e22821, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34036678

ABSTRACT

The preservation of the redox homeostasis is critical for cell survival and functionality. Redox imbalance is an essential inducer of several pathological states. CD4+ /helper T cells are highly dependent on the redox state of their surrounding milieu. The potential of the aryl hydrocarbon receptor (AhR) engagement in controlling CD4+ T-cell fate during redox alteration is still challenging. C57BL/6 mice were treated with AhR agonist 6-formylindolo[3,2-b]carbazole (FICZ), AhR antagonist CH223191, an inhibitor of glutathione biosynthesis buthionine sulfoximine (BSO), and the antioxidant N-acetylcysteine (NAC) alone or in combination. Six days later, splenocytes were evaluated for the expression of the redox-related genes and the possible changes in T-cell subsets. FICZ like BSO significantly elevated the expression of HMOX1, GCLC, and GCLM genes but it failed to increase the expression of the Nrf2 gene. Moreover, FICZ + BSO increased while FICZ + CH223191 or NAC decreased the expression of these genes. FICZ also significantly increased Th1 cell numbers but decreased Tregs in a dose-dependent manner. Furthermore, a high dose of FICZ + CH223191 + NAC significantly enhanced Th1, Th17, and Treg cells but its low dose in such a situation increased Th2 and Th17 while decreased Treg cells. AhR engagement during redox alteration can determine the fate of CD4 + T cells, so, AhR agonists or antagonists might be useful in assessing immune responses. However, these results need further verifications in vitro and in animal models of various diseases.


Subject(s)
Receptors, Aryl Hydrocarbon , T-Lymphocytes, Helper-Inducer/metabolism , Acetylcysteine/pharmacology , Animals , Azo Compounds/pharmacology , Gene Expression Regulation/drug effects , Glutamate-Cysteine Ligase/biosynthesis , Heme Oxygenase-1/biosynthesis , Membrane Proteins/biosynthesis , Mice , NF-E2-Related Factor 2/biosynthesis , Oxidation-Reduction/drug effects , Pyrazoles/pharmacology , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/metabolism
4.
Neurotoxicology ; 84: 53-63, 2021 05.
Article in English | MEDLINE | ID: mdl-33617922

ABSTRACT

Alzheimer's disease (AD) etiology has been studied for a long time and it is found to be multifaceted involving the accumulation of amyloid ß and tau protein. Oxidative stress is an early event in AD associated neurodegeneration provoking neuronal death through mitochondrial dysfunction and activation of caspase-3. Therefore we tested the efficacy of dihydroactinidiolide (DHAc), a monoterpene lactone against the oxidative load involved in AD like pathological conditions induced by sodium dithionite, glutamate, amyloid ß and colchicine in SH-SY5Y cells. Some of the indicators of neurotoxicity like acetylcholinesterase activity, intracellular reactive oxygen species (ROS), nitrite content, lipid peroxidation, protein carbonylation, nuclear and membrane damage were found to be significantly high in the toxicant treated cells when compared to the control cells while DHAc pretreatment significantly restored the toxicant induced neuronal damage signatures. Caspase-3 activity was found to be increased in the toxicant treated cells while DHAc significantly reduced it. Western blotting and RT-PCR revealed that DHAc significantly increased anti-apoptotic Bcl-2 expression and mRNA levels of Nrf2 and HO-1. Therefore DHAc was found to protect SH-SY5Y cells from neurotoxicant induced oxidative stress and apoptosis by regulating cellular antioxidant defenses and apoptosis related genes.


Subject(s)
Benzofurans/pharmacology , Caspase Inhibitors/pharmacology , Heme Oxygenase-1/biosynthesis , NF-E2-Related Factor 2/biosynthesis , Neuroblastoma/metabolism , Oxidative Stress/drug effects , bcl-2-Associated X Protein/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/biosynthesis , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/physiology , bcl-2-Associated X Protein/biosynthesis
5.
Int J Mol Sci ; 22(3)2021 Feb 02.
Article in English | MEDLINE | ID: mdl-33540888

ABSTRACT

Macrophages are essential immune cells of the innate immune system. They participate in the development and regulation of inflammation. Macrophages play a fundamental role in fighting against bacterial infections by phagocytosis of bacteria, and they also have a specific role in immunomodulation by secreting pro-inflammatory cytokines. In bacterial infection, macrophages decrease the serum iron concentration by removing iron from the blood, acting as one of the most important regulatory cells of iron homeostasis. We examined whether the Gram-positive and Gram-negative cell wall components from various bacterial strains affect the cytokine production and iron transport, storage and utilization of THP-1 monocytes in different ways. We found that S. aureus lipoteichoic acid (LTA) was less effective in activating pro-inflammatory cytokine expression that may related to its effect on fractalkine production. LTA-treated cells increased iron uptake through divalent metal transporter-1, but did not elevate the expression of cytosolic and mitochondrial iron storage proteins, suggesting that the cells maintained iron efflux via the ferroportin iron exporter. E. coli and P. aeruginosa lipopolysaccharides (LPSs) acted similarly on THP-1 cells, but the rates of the alterations of the examined proteins were different. E. coli LPS was more effective in increasing the pro-inflammatory cytokine production, meanwhile it caused less dramatic alterations in iron metabolism. P. aeruginosa LPS-treated cells produced a smaller amount of pro-inflammatory cytokines, but caused remarkable elevation of both cytosolic and mitochondrial iron storage proteins and intracellular iron content compared to E. coli LPS. These results prove that LPS molecules from different bacterial sources alter diverse molecular mechanisms in macrophages that prepossess the outcome of the bacterial infection.


Subject(s)
Cell Wall/chemistry , Cytokines/metabolism , Escherichia coli/chemistry , Iron/metabolism , Lipopolysaccharides/pharmacology , Pseudomonas aeruginosa/chemistry , Staphylococcus aureus/chemistry , THP-1 Cells/metabolism , Teichoic Acids/pharmacology , Biological Transport , CX3C Chemokine Receptor 1/biosynthesis , CX3C Chemokine Receptor 1/genetics , Chemokine CX3CL1/metabolism , Cytokines/biosynthesis , Cytosol/metabolism , Ferritins/biosynthesis , Ferritins/genetics , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Hepcidins/biosynthesis , Hepcidins/genetics , Humans , Mitochondria/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oxidoreductases/biosynthesis , Oxidoreductases/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/genetics , THP-1 Cells/drug effects
6.
Mol Med Rep ; 23(3)2021 03.
Article in English | MEDLINE | ID: mdl-33398364

ABSTRACT

Isoegomaketone (IK) is a known component of Perilla frutescens that reportedly exhibits anti­inflammatory, anti­cancer and anti­allergic properties. A novel compound known as 9­HIK has been isolated from the extract of a radiation mutant P. frutescens var. crispa using supercritical carbon dioxide. In the present study, 9­HIK induced heme oxygenase­1 (HO­1) mRNA expression in RAW264.7 cells, with maximal levels observed 4 h after 9­HIK treatment. In addition, 9­HIK inhibited the mRNA and protein expression of pro­inflammatory mediators, such as IL­6 and interferon­ß, as well as the production of nitric oxide (NO) in lipopolysaccharide­stimulated RAW264.7 cells. Furthermore, N­acetyl­L­cysteine, a reactive oxygen species scavenger, inhibited NO production and HO­1 mRNA expression levels through the nuclear factor erythroid 2­related factor 2 pathway. Overall, 9­HIK displayed anti­inflammatory properties in LPS­induced RAW264.7 cells via direct suppression of inflammatory mediators and HO­1 induction.


Subject(s)
Furans/pharmacology , Interferon-beta/biosynthesis , Interleukin-6/biosynthesis , Ketones/pharmacology , Lipopolysaccharides/toxicity , Nitric Oxide/metabolism , Animals , Furans/chemistry , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/biosynthesis , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Ketones/chemistry , Membrane Proteins/biosynthesis , Mice , RAW 264.7 Cells
7.
Oxid Med Cell Longev ; 2020: 1080168, 2020.
Article in English | MEDLINE | ID: mdl-33343802

ABSTRACT

Heme oxygenase-1 (HO-1) has been shown to exert as an antioxidant and anti-inflammatory enzyme in cardiovascular inflammatory diseases. Flavonoids have been demonstrated to display anti-inflammatory and antioxidant effects through the induction of HO-1. 5,8-Dihydroxy-4',7-dimethoxyflavone (DDF), one of the flavonoid compounds, is isolated from Reevesia formosana. Whether DDF induced HO-1 expression on human cardiac fibroblasts (HCFs) remained unknown. Here, we found that DDF time- and concentration-dependently induced HO-1 protein and mRNA expression, which was attenuated by pretreatment with reactive oxygen species (ROS) scavenger N-acetyl cysteine (NAC) in HCFs. DDF-enhanced ROS generation was attenuated by NAC, but not by either diphenyleneiodonium chloride (DPI, Nox inhibitor) or MitoTempol (mitochondrial ROS scavenger). Interestingly, pretreatment with glutathione (GSH) inhibited DDF-induced HO-1 expression. The ratio of GSH/GSSG was time-dependently decreased in DDF-treated HCFs. DDF-induced HO-1 expression was attenuated by an inhibitor of p38 MAPK (p38i VIII) or siRNA, but not by MEK1/2 (PD98059) or JNK1/2 (SP600125). DDF-stimulated p38 MAPK phosphorylation was inhibited by GSH or p38i VIII. Moreover, DDF-induced HO-1 expression was mediated through Nrf2 phosphorylation and translocation into the nucleus which was attenuated by NAC or p38 siRNA. DDF also stimulated antioxidant response element (ARE) promoter activity which was inhibited by NAC, GSH, or p38i VIII. Interaction between Nrf2 and the ARE-binding sites on the HO-1 promoter was revealed by chromatin immunoprecipitation assay, which was attenuated by NAC, GSH, or p38i VIII. We further evaluated the functional effect of HO-1 expression on the thrombin-induced fibrotic responses. Our result indicated that the induction of HO-1 by DDF can attenuate the thrombin-induced connective tissue growth factor expression. These results suggested that DDF-induced HO-1 expression is, at least, mediated through the activation of the ROS-dependent p38 MAPK/Nrf2 signaling pathway in HCFs. Thus, the upregulation of HO-1 by DDF could be a candidate for the treatment of heart fibrosis.


Subject(s)
Connective Tissue Growth Factor/biosynthesis , Fibroblasts/metabolism , Flavones/pharmacology , Heme Oxygenase-1/biosynthesis , MAP Kinase Signaling System/drug effects , Myocardium/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Thrombin/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Line , Connective Tissue Growth Factor/genetics , Enzyme Induction/drug effects , Heme Oxygenase-1/genetics , Humans , NF-E2-Related Factor 2/genetics , p38 Mitogen-Activated Protein Kinases/genetics
8.
Int J Mol Sci ; 21(22)2020 Nov 14.
Article in English | MEDLINE | ID: mdl-33202535

ABSTRACT

Ultraviolet B (UV-B) radiation induces the extreme production of either reactive oxygen species (ROS) or inflammatory mediators. The aim of this study was to evaluate the antioxidant activities of 70% ethanolic extract of Lablab purpureus (LPE) and the underlying mechanisms using HaCaT cells exposed to UV-B. High-performance liquid chromatography (HPLC) confirmed the presence of gallic acid, catechin, and epicatechin in LPE. LPE was shown to have a very potent capacity to scavenge free radicals. The results showed that LPE prevented DNA damage and inhibited the generation of ROS in HaCaT cells without causing any toxicity. LPE increased the expression of endogenous antioxidant enzymes such as superoxide dismutase-1 and catalase. Furthermore, LPE treatment facilitates the nuclear translocation of nuclear factor (erythroid-derived 2)-like 2 (Nrf-2), boosting the phase II detoxifying enzyme heme oxygenase-1 (HO-1) leading to the combatting of oxidative stress. However, pretreatment of LPE also caused the phosphorylation of mitogen-activated protein kinases (MAPK kinase) (p38 kinase) and extracellular signal-regulated kinase (ERK), whereas treatment with p38 and ERK inhibitors substantially suppressed LPE-induced Nrf2 and heme oxygenase (HO)-1 expression. These findings suggest that LPE exhibits antioxidant activity via Nrf-2-mediated HO-1 signaling through the activation of p38 and ERK, indicating that LPE can potentially be used as a remedy to combat oxidative stress-induced disorder.


Subject(s)
Fabaceae/chemistry , Free Radical Scavengers/pharmacology , Heme Oxygenase-1/biosynthesis , Keratinocytes/metabolism , MAP Kinase Signaling System , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Plant Extracts , Ultraviolet Rays/adverse effects , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Line , Free Radical Scavengers/chemistry , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/radiation effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Plant Extracts/chemistry , Plant Extracts/pharmacology
9.
Cell Rep ; 33(4): 108317, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33113362

ABSTRACT

Hematophagous vectors lacerate host skin and capillaries to acquire a blood meal, resulting in leakage of red blood cells (RBCs) and inflammation. Here, we show that heme oxygenase-1 (HO-1), a pleiotropic cytoprotective isoenzyme that mitigates heme-mediated tissue damage, is induced after bites of sand flies, mosquitoes, and ticks. Further, we demonstrate that erythrophagocytosis by macrophages, including a skin-residing CD163+CD91+ professional iron-recycling subpopulation, produces HO-1 after bites. Importantly, we establish that global deletion or transient inhibition of HO-1 in mice increases inflammation and pathology following Leishmania-infected sand fly bites without affecting parasite number, whereas CO, an end product of the HO-1 enzymatic reaction, suppresses skin inflammation. This indicates that HO-1 induction by blood-feeding sand flies promotes tolerance to Leishmania infection. Collectively, our data demonstrate that HO-1 induction through erythrophagocytosis is a universal mechanism that regulates skin inflammation following blood feeding by arthropods, thus promoting early-stage disease tolerance to vector-borne pathogens.


Subject(s)
Dermatitis/enzymology , Heme Oxygenase-1/biosynthesis , Insect Bites and Stings/enzymology , Vector Borne Diseases/enzymology , Vector Borne Diseases/pathology , Animals , Arthropods , Culicidae , Dermatitis/pathology , Female , Insect Bites and Stings/pathology , Leishmania , Leishmaniasis/enzymology , Mice , Mice, Inbred C57BL
10.
Postgrad Med ; 132(8): 720-726, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32757691

ABSTRACT

OBJECTIVES: This study aimed to investigate the relationships among islet function, the Nrf2 pathway, and insulin receptor substrate 2 (IRS2) in type 2 diabetes mellitus (T2DM), prediabetes mellitus (IGR), and normal glucose tolerance (NGT) populations. METHODS: Three hundred cases each were selected for the NGT, IGR, and T2DM groups; FBG, 2hPG, HbA1 c, FINS, TG, TC, HDL-C, and LDL-C levels and serum levels of nuclear factor in E2-related factor 2 (Nrf2), insulin receptor substrate 2 (IRS2), tumor necrosis factor alpha (TNF-α), and heme oxygenase 1 (HO-1) were evaluated. RESULTS: The T2DM group had lower islet ß-cell function index and insulin sensitivity index than the NGT and IGR groups (P < 0.05). The Nrf2, IRS2, and HO-1 levels in the NGT, IGR, and T2DM groups followed a decreasing trend in the order mentioned, whereas the TNF-α levels followed an increasing trend. CONCLUSIONS: Upon impairment of glucose regulation, the expression of TNF-α in the human body increased, which indicated the aggravation of oxidative stress (OS) and the inflammatory response. Islet function was maintained in the pre-diabetic population, and concurrently, the TNF-α, Nrf2, and HO-1 levels were moderately elevated, the expression of IRS2 was marginally inhibited, and the Nrf2 pathway was activated under mild OS stimulus to resist OS, inflammation, and injury, which may have been mediated through PI3 K/AKT. In patients with T2DM, islet function was significantly poorer, TNF-α amplification was enhanced significantly, and Nrf2, HO-1, and IRS2 expression reduced significantly; this suggested that, along with the aggravation of OS and the inflammatory response, Nrf2 pathway activation and HO-1 expression were both inhibited, the antioxidant capacity of the body was reduced, IRS2 degradation increased, and islet function was impaired.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Insulin Receptor Substrate Proteins/metabolism , Insulin-Secreting Cells/metabolism , NF-E2-Related Factor 2/metabolism , Aged , Blood Glucose , Blood Pressure , Body Weights and Measures , Cross-Sectional Studies , Female , Glycated Hemoglobin , Heme Oxygenase-1/biosynthesis , Humans , Lipids/blood , Male , Middle Aged , Prediabetic State/physiopathology , Tumor Necrosis Factor-alpha/biosynthesis
11.
Biomed Pharmacother ; 130: 110553, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32739742

ABSTRACT

1) BACKGROUND: Phycocyanin (PC) is a type of natural protein in algae with antioxidant and anti-inflammatory properties. However, the protective effect of PC on hepatic damage induced by X-ray remains unknown. 2) METHODS: Male C57BL/6 mice were gavaged with 200mg/kg PC for consecutive 7 days before or after radiation. The blood samples and tissues were collected on days 1 and 7 after radiation exposure. 3) RESULTS: Pretreatment or treatment with PC decreased significantly the levels of alanine aminotransferase (ALT), aspartate aminotransferase(AST) in the plasma. Histological evaluation further confirmed the protection of PC against radiation-induced hepatotoxicity. PC-treatment also increased the relative mRNA expression of superoxide dismutase (SOD) and glutathione (GSH-PX), and descended the ROS in the liver. Moreover, the expression of H2AX, an indicator of DNA damage in mice, of the PC-intervention group was much smaller than that of the radiation group. In vivo, PC-treatment markedly up-regulated NF-E2-related factor 2(Nrf2) expression and downstream gene such as hemeoxygenase-1 (HO-1), NQO1. 4) Conclusion: PC could attenuate the radiation-induced oxidative stress damage by activating Nrf2/ HO-1 signaling pathway, and reduce the radiation-induced DNA damage. Therefore, PC is a protective agent against radiation-induced liver damage.


Subject(s)
Liver Diseases/drug therapy , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Phycocyanin/therapeutic use , Radiation Injuries, Experimental/drug therapy , X-Rays , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Glutathione Peroxidase/metabolism , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Reactive Oxygen Species/metabolism , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/drug effects
12.
Neuroreport ; 31(15): 1065-1071, 2020 10 14.
Article in English | MEDLINE | ID: mdl-32804709

ABSTRACT

BACKGROUND: A growing body of experimental evidence suggests that hemin released from heme is a potent oxidant and accumulates in intracranial hematomas. Hemopexin (Hpx) decreases hemin accumulation and catabolism by nerve cells. In previous study, we observed that Hpx gene knockout aggravated striatal injury and worsened behavioral deficits of mice subjected to intracerebral hemorrhage. AIM: To examine the effect of Hpx on oxidative damage and apoptosis in cultured nerve cells with blood clot. METHODS: Neuron and glial cells were transfected with adenoviral Hpx gene. Transfected primary neuron-glial cells were co-cultured with 50 µl of arterial blood clot using insert transwells. The sham group was co-coulture with 50 µl of DMEM/F12, which contained 28 µl of serum; the control group was transfected with adenoviral vector. At 12 and 24 h, the level of malonaldehyde (MDA), surperoxide dismutase (SOD) concentration, glutathione (GSH), apoptosis, expression of HO-1 and caspase-3 were detected. RESULTS: MDA level was decreased (P < 0.01) whereas SOD and GSH concentration were increased in the Hpx group (P < 0.05 and P < 0.01, respectively). Results of flow cytometry revealed no significant difference in apoptosis between the Hpx group and model group at 12 h. However, the percentage of cells undergoing apoptosis in the Hpx group was decreased at 24 h compared with the model group (P < 0.01). HO-1 expression decreased in the Hpx group at 24 h (P < 0.01) while caspase-3 expression decreased at both 12 and 24 h (P < 0.011 and P < 0.05, respectively) compared with the model group. CONCLUSION: Hpx protected nerve cells exposed to blood from injury by anti-oxidation and a decrease in the expression of HO-1 and caspase-3.


Subject(s)
Adenoviridae , Apoptosis/physiology , Cerebral Cortex/metabolism , Hemopexin/biosynthesis , Oxidative Stress/physiology , Thrombosis/metabolism , Adenoviridae/genetics , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/pathology , Coculture Techniques , Gene Transfer Techniques , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Hemopexin/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Neuroglia/metabolism , Neuroglia/pathology , Rats , Rats, Sprague-Dawley , Thrombosis/pathology , Thrombosis/prevention & control
13.
J Nat Prod ; 83(9): 2647-2654, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32816473

ABSTRACT

Eight new dioxopiperazine alkaloids, penispirozines A-H (1-8), were discovered from the mangrove-derived fungus Penicillium janthinellum HDN13-309. Their structures were elucidated by spectroscopic analysis, TDDFT-ECD calculations, and X-ray diffraction. Compound 1 had an unusual pyrazino[1,2]oxazadecaline coupled with a thiophane ring system, and compound 2 possessed a 6/5/6/5/6 pentacyclic ring system with two rare spirocyclic centers. Interestingly, compounds 3-8 were distinguished by not only the existence of a spiro-thiophane or spiro-furan ring system but also the chirality of the pentacyclic moiety. Compounds 3 and 4 increased the expression of the two relevant phase II detoxifying enzymes SOD2 and HO-1 at 10 µM.


Subject(s)
Alkaloids/chemistry , Avicennia/microbiology , Penicillium/chemistry , Crystallography, X-Ray , Enzyme Induction/drug effects , Fermentation , Heme Oxygenase-1/biosynthesis , Magnetic Resonance Spectroscopy , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Superoxide Dismutase/biosynthesis , X-Ray Diffraction
14.
Molecules ; 25(14)2020 Jul 08.
Article in English | MEDLINE | ID: mdl-32650607

ABSTRACT

Activation of nuclear factor erythroid-2-related factor 2 (NRF2) has been proven to be an effective means to prevent the development of cancer, and natural curcumin stands out as a potent NRF2 activator and cancer chemopreventive agent. In this study, we have synthesized a series of 4-anilinoquinolinylchalcone derivatives, and used a NRF2 promoter-driven firefly luciferase reporter stable cell line, the HaCaT/ARE cells, to screen a panel of these compounds. Among them, (E)-3-{4-[(4-acetylphenyl)amino]quinolin-2-yl}-1-(4-fluorophenyl)prop-2-en-1-one (13b) significantly increased NRF2 activity in the HaCaT cell with a half maximal effective concentration (EC50) value of 1.95 µM. Treatment of compound 13b upregulated HaCaT cell NRF2 expression at the protein level. Moreover, the mRNA level of NRF2 target genes, heme oxygenase-1 (HO-1), glutamate-cysteine ligase catalytic subunit (GCLC), and glucose-6-phosphate dehydrogenase (G6PD) were significantly increased in HaCaT cells upon the compound 13b treatment. The molecular docking results exhibited that the small molecule 13b is well accommodated by the bound region of Kelch-like ECH-associated protein 1 (Keap1)-Kelch and NRF2 through stable hydrogen bonds and hydrophobic interaction, which contributed to the enhancement of affinity and stability between the ligand and receptor. Compound 13b has been identified as the lead compound for further structural optimization.


Subject(s)
Chalcones , Gene Expression Regulation/drug effects , Kelch-Like ECH-Associated Protein 1 , Keratinocytes , Molecular Docking Simulation , NF-E2-Related Factor 2/biosynthesis , Cell Line, Transformed , Chalcones/chemical synthesis , Chalcones/chemistry , Chalcones/pharmacology , Glucosephosphate Dehydrogenase , Glutamate-Cysteine Ligase/biosynthesis , Heme Oxygenase-1/biosynthesis , Humans , Kelch-Like ECH-Associated Protein 1/chemistry , Kelch-Like ECH-Associated Protein 1/metabolism , Keratinocytes/chemistry , Keratinocytes/metabolism , NF-E2-Related Factor 2/genetics
15.
J Alzheimers Dis ; 76(2): 623-641, 2020.
Article in English | MEDLINE | ID: mdl-32568195

ABSTRACT

BACKGROUND: Adult hippocampal neurogenesis is critical for renewing hippocampal neural circuits and maintaining hippocampal cognitive function and is closely associated with age-related neurodegenerative diseases. Heme oxygenase 1 (HO-1) is a stress protein that catalyzes the degradation of heme into free iron, biliverdin, and carbon monoxide. Elevated HO-1 level constitutes a pathological feature of Alzheimer's disease, Parkinson's disease, and many other age-related neurodegenerative diseases. OBJECTIVE: Here we research the precise role of HO-1 in adult hippocampal neurogenesis. METHODS: To explore the effect of HO-1 overexpression on adult neural stem cells (aNSCs) and elucidate its mechanisms, Tg(HO-1) was constructed. The transgenic mice and aNSCs were subjected to neurosphereing assay, clonal analysis, and BrdU labelling to detect the proliferation and self-renewal ability. LiCl, MG132, CHX, and IGF-1 treatment were used to research the signaling pathways which regulated by HO-1. RESULTS: HO-1 overexpression decreased proliferation ability and induced apoptosis of aNSCs in subgranular zoon (SGZ) in vivo and in vitro. Furthermore, HO-1 overexpression inactivated canonical WNT/ß-catenin pathway. Re-activate canonical WNT/ß-catenin pathway rescued aNSCs proliferation and survival upon HO-1 overexpression. More importantly, phosphorylation of AKTS473 and GSK3ßS9 was found to be significantly decreased in HO-1 overexpressed aNSCs. Re-activation of AKT signaling proved that HO-1 inhibited Wnt/ß-catenin signaling pathway via AKT/GSK3ß signaling pathway. CONCLUSION: These results demonstrated a critical role of HO-1 in regulating aNSCs survival and proliferation by inhibiting Wnt/ß-catenin pathway through repression of AKT/GSK3ß, which provide a novel insight into the role of HO-1 in Alzheimer's disease pathogenesis.


Subject(s)
Cell Proliferation/physiology , Heme Oxygenase-1/biosynthesis , Membrane Proteins/biosynthesis , Neural Stem Cells/metabolism , Neurogenesis/physiology , Wnt Signaling Pathway/physiology , Age Factors , Animals , Cell Survival/physiology , Cells, Cultured , Female , Hippocampus/cytology , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
16.
Immunopharmacol Immunotoxicol ; 42(4): 295-305, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32397768

ABSTRACT

AIM: Acute rejection is still a major problem in transplantation and one of the most important causes of late graft loss. Cyclosporine and tacrolimus are widely used for suppression of T cell function to avoid graft rejection, but long-term use of these compounds is associated with serious toxicities. Quercetin, a flavonoid found in fruits and vegetables, has been demonstrated to exhibit cytoprotective effects through the induction of heme oxygenase (HO) -1, an enzyme involved in heme catabolism. We hypothesized that quercetin induces HO-1 in T cells and suppresses T cell function via HO-1. In the present study, we showed that quercetin suppressed the A23187-mediated expression of interleukin (IL) -2 in T cells. METHODS: Mouse splenocytes, enriched T cells, and EL4 cells, a mouse T cell line, were treated with quercetin, and then stimulated with A23187, a calcium ionophore, concanavalin A, or anti-CD3ε and anti-CD28 antibodies. Cell proliferation, expression of IL-2, calcium mobilization, apoptosis, cell cycle, and phosphorylation of extracellular signal-regulated kinase (ERK) were investigated. RESULTS: Quercetin induced HO-1, and this induction of HO-1 was implicated in the suppression of IL-2 production. Furthermore, the induction of HO-1 by quercetin suppressed the influx of calcium ions, a known trigger of IL-2 production. Additionally, quercetin suppressed T cell proliferation through promotion of cell cycle arrest via HO-1 induction, but quercetin did not induce apoptosis. To investigate the role of the signal transduction pathway in quercetin's effect on cell proliferation, we evaluated the phosphorylation of ERK in T cells. Quercetin suppressed the A23187-mediated stimulation of ERK, an effect that was mediated through HO-1. These results suggested that HO-1 is involved in the suppressive effects of quercetin on T cell activation and proliferation. CONCLUSION: Our findings indicate that the quercetin may be a promising candidate for inducing HO-1 in T cells, thereby facilitating immunosuppressive effects.


Subject(s)
Antioxidants/pharmacology , Heme Oxygenase-1/biosynthesis , Quercetin/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , Enzyme Induction/physiology , Mice , Mice, Inbred C57BL
17.
Int Immunopharmacol ; 83: 106437, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32222637

ABSTRACT

In this study, we investigated the renoprotective effects and mechanism of isovitexin, a glycosylflavonoid isolated from rice hulls of Oryza sativa, against cisplatin-induced kidney injury in mice. The mice were treated with cisplatin for four consecutive days and at the second day, the mice were received with isovitexin for three consecutive days. The levels of blood urea nitrogen (BUN) and creatinine in serum and the levels of MDA, ROS, TNF-α, IL-1ß and IL-6 in kidney tissues were measured. The proteins of Nrf2 and NF-κB signaling pathways were measured by western blot analysis. Our results demonstrated that isovitexin inhibited CP-induced increases in serum BUN and creatinine. Isovitexin inhibited CP-induced inflammation by inhibiting TNF-α, IL-1ß and IL-6 production in kidney tissues. Also, isovitexin inhibited CP-induced oxidative stress by inhibiting MDA and ROS production. Furthermore, isovitexin was found to inhibit CP-induced NF-κB activation and increase Nrf2 and HO-1 expression. In conclusion, our results indicated that isovitexin protected against CP-induced kidney injury by suppressing inflammatory and oxidative responses.


Subject(s)
Acute Kidney Injury/prevention & control , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apigenin/pharmacology , Inflammation/prevention & control , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Apigenin/therapeutic use , Blood Urea Nitrogen , Cisplatin/toxicity , Creatinine/blood , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Female , Heme Oxygenase-1/biosynthesis , Malondialdehyde/metabolism , Mice , Mice, Inbred BALB C , NF-E2-Related Factor 2/biosynthesis , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
18.
Biomolecules ; 10(3)2020 03 01.
Article in English | MEDLINE | ID: mdl-32121588

ABSTRACT

Mevastatin (MVS) has been previously shown to induce heme oxygenase (HO)-1 expression through Nox/ROS-dependent PDGFRα/PI3K/Akt/Nrf2/ARE axis in human pulmonary alveolar epithelial cells (HPAEpiCs). However, alternative signaling pathways might involve in MVS-induced HO-1 expression. We found that tumor necrosis factor α (TNFα) induced vascular cell adhesion protein 1 (VCAM-1) expression and NF-κB p65 phosphorylation which were attenuated by pretreatment with MVS via up-regulation of HO-1, determined by Western blot and real-time qPCR. TNFα-induced VCAM-1 expression was attenuated by an NF-κB inhibitor, Bay117082. The inhibitory effects of MVS were reversed by tin protoporphyrin (SnPP)IX (an inhibitor of HO-1 activity). In addition, pretreatment with the inhibitor of pan-Protein kinase C (PKC) (GF109203X), PKCα (Gö6983), Pyk2 (PF431396), p38α MAPK (SB202190), JNK1/2 (SP600125), or AP-1 (Tanshinone IIA), and transfection with their respective siRNAs abolished MVS-induced HO-1 expression in HPAEpiCs. c-Jun (one of AP-1 subunits) was activated by PKCα, Pyk2, p38α MAPK, and JNK1/2, which turned on the transcription of the homx1 gene. The interaction between c-Jun and HO-1 promoter was confirmed by a chromatin immunoprecipitation (ChIP) assay, which was attenuated by these pharmacological inhibitors. These results suggested that MVS induces AP-1/HO-1 expression via PKCα/Pyk2/p38α MAPK- or JNK1/2-dependent c-Jun activation, which further binds with AP-1-binding site on HO-1 promoter and suppresses the TNFα-mediated inflammatory responses in HPAEpiCs. Thus, upregulation of the AP-1/HO-1 system by MVS exerts a potentially therapeutic strategy to protect against pulmonary inflammation.


Subject(s)
Alveolar Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Heme Oxygenase-1/biosynthesis , Lovastatin/analogs & derivatives , Monocytes/metabolism , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Cell Adhesion/drug effects , Cell Line , Humans , Lovastatin/pharmacology
19.
J Ethnopharmacol ; 254: 112681, 2020 May 23.
Article in English | MEDLINE | ID: mdl-32087320

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Antrodia camphorata (A. camphorata) is a rare functional fungus in Taiwan and contains a variety of biologically active ingredients. Antrodin A (AdA) is one of the main active ingredients in the solid-state fermented A. camphorata mycelium. It protects the liver from alcohol damage by improving the antioxidant and anti-inflammatory capacity of the liver and maintaining the stability of the intestinal flora. AIM OF THE STUDY: The aim of this study was to evaluate the hepatoprotective activities of ethyl acetate layer extract (EALE), AdA, and Antroquinonol (Aq) from mycelium of A. camphorata on alcoholic liver injury. MATERIALS AND METHODS: Mice were given with intragastrically vehicle (NC, 2% CMC-Na), alcohol (AL, 12 mL/kg bw), or different A. camphorata samples (EALE, AdA, Aq) at low (100 mg/kg bw) or high (200 mg/kg bw) dosages. The positive control (PC) group was given with silymarin (200 mg/kg bw). Except the NC group, each group of mice was fasted for 4 h after the last treatment and was intragastrically administrated with 50% alcohol (12 mL/kg bw). At the end of experiment, mouse serum was collected and the liver was excised. A portion of the liver was fixed in formalin and used for histopathological analysis, whereas the rest was used for biochemical analysis and real-time PCR analysis. The intestinal flora structure of feces was analyzed by determining the v3-v4 region sequence in 16S rDNA. RESULTS: The high-dose groups of the three samples (EALEH, AdAH, and AqH) significantly alleviated the alcohol-induced increases in liver index, serum ALT, AST, and AKP activities. Serum TG level was significantly reduced in all treatment groups. The increase of HDL-C content indicated that active ingredients of A. camphorata could reduce the lipid content in serum. Furthermore, MDA contents of the AdAH and AqH groups in liver were significantly reduced, accompanying with the levels of SOD, CAT, and GSH elevated to various extents. Antioxidant and anti-inflammatory capabilities in the liver were increased in the AdAH group, as evidenced by the mRNA expression levels of Nrf-2 and HO-1 were significantly increased; while those of CYP2e1, TNF-α, and TLR-4 were significantly decreased. Analysis of intestinal flora of feces showed that alcohol treatment significantly changed the composition of intestinal flora. Supplementation with AdA could mitigate dysbiosis of intestinal flora induced by alcohol. Flora of Faecalibaculum, Lactobacillus, and Coriobacteriaceae_UCG-002 showed significantly negative correlations with ALT, AST, AKP, and MDA levels. CONCLUSION: Antrodin A could improve the antioxidant and anti-inflammatory capacities of the liver and maintain the stability of intestinal flora. It is potentially a good candidate compound against acute alcoholic liver injury.


Subject(s)
Antrodia/chemistry , Dysbiosis/prevention & control , Liver Diseases, Alcoholic/prevention & control , Maleic Anhydrides/pharmacology , Animals , Complex Mixtures/pharmacology , Cytochrome P-450 CYP2E1/biosynthesis , Heme Oxygenase-1/biosynthesis , Intestines/microbiology , Liver/metabolism , Liver Function Tests , Male , Membrane Proteins/biosynthesis , Mice , Microbiota/drug effects , Mycelium/chemistry , NF-E2-Related Factor 2/biosynthesis , Protective Agents/pharmacology , Silymarin/pharmacology , Toll-Like Receptor 4/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology
20.
Mol Neurobiol ; 57(5): 2436-2446, 2020 May.
Article in English | MEDLINE | ID: mdl-32108290

ABSTRACT

In human glioma tumours, heme oxygenase-1 (HO-1) is overexpressed when compared with normal brain tissues and during oligodendroglioma progression. However, the molecular mechanisms mediated by HO-1 to promote glioblastoma remain unknown. We therefore aimed at investigating the effect of HO-1 expression and its selective enzymatic inhibition in two different cell lines (i.e. A172 and U87-MG). HO-1 was induced by hemin treatment (10 µM), and VP13/47 (100 µM) was used as a specific non-competitive inhibitor of HO-1 activity. Cell proliferation was measured by cell index measurement (xCelligence technology) and clonogenic assay, whereas cell migration was assessed by wound healing assay. Carbon monoxide-releasing molecules (CORMs) (i.e. CORM-3 and CORM-A1) were also used in a separate set of experiments to confirm the effect of HO-1 by-product in glioblastoma progression further. Our results were further validated using GSE4412 microarray dataset analysis and comparing biopsies overexpressing HO-1 with the rest of the cases. Our results showed that hemin was able to induce both HO-1 gene and protein expression in a cell-dependent manner being A172 more responsive to pharmacological upregulation of HO-1. Hemin, but not CORMs treatment, resulted in a significant increase of cell proliferation following 24 h of treatment as measured by increased cell index and colony formation capacity and such effect was abolished by VP13/47. Interestingly, both hemin and CORMs showed a significant effect on the wound healing assay also exhibiting cell specificity. Finally, our dataset analysis showed a positive correlation of HO-1 gene expression with ITGBI and ITGBII which are membrane receptors involved in cell adhesion, embryogenesis, tissue repair, immune response and metastatic diffusion of tumour cells. In conclusion, our data suggest that HO-1 and its by-product CO exhibit a cell-specific effect on various aspects of disease progression and are associated with a complex series of molecular mechanisms driving cell proliferation, survival and metastasis.


Subject(s)
Brain Neoplasms/pathology , Carbon Monoxide/physiology , Glioblastoma/pathology , Heme Oxygenase-1/physiology , Neoplasm Proteins/physiology , Boranes/pharmacology , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Carbonates/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Chemotaxis/drug effects , Datasets as Topic , Disease Progression , Enzyme Induction/drug effects , Gene Expression Profiling , Gene Ontology , Glioblastoma/enzymology , Glioblastoma/genetics , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Hemin/pharmacology , Humans , Hydrocarbons, Brominated/pharmacology , Imidazoles/pharmacology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Organometallic Compounds/pharmacology , Tumor Stem Cell Assay
SELECTION OF CITATIONS
SEARCH DETAIL
...